Regulation of the Stability of the Protein Kinase DYRK1A: Establishing Connections with the Wnt Signaling Pathway

Total Page:16

File Type:pdf, Size:1020Kb

Regulation of the Stability of the Protein Kinase DYRK1A: Establishing Connections with the Wnt Signaling Pathway Regulation of the stability of the protein kinase DYRK1A: establishing connections with the Wnt signaling pathway Krisztina Arató TESI DOCTORAL UPF / 2010 Barcelona, November 2010 Regulation of the stability of the protein kinase DYRK1A: establishing connections with the Wnt signaling pathway Krisztina Arató Memòria presentada per optar al grau de Doctora per la Universitat Pompeu Fabra. Aquesta tesi ha estat realitzada sota la direcció de la Dra. Susana de la Luna al Centre de Regulació Genòmica (CRG, Barcelona), dins del Programa de Genes i Malaltia. Krisztina Arató Susana de la Luna A Pere, por haberme traído a Barcelona… Cover design by Luisa Lente (www.yoyo.es). Index Page Abstract/Resumen.................................................................................. 1 Introduction............................................................................................. 5 The protein kinase DYRK1A................................................................ 7 The DYRK family of protein ....................................................... 7 Structure and mechanism of activation of DYRK1A kinase........ 8 Regulation of DYRK1A expression ............................................ 10 Regulation of DYRK1A subcellular localization.......................... 11 Regulation of DYRK1A activity................................................... 13 DYRK1A as a regulator of signaling pathways........................... 14 The Notch signaling pathway........................................... 16 Receptor tyrosine kinase signaling.................................. 17 Hedgehog signaling......................................................... 19 Calcineurin-NFAT signaling............................................. 20 DYRK1A and disease................................................................. 22 Nemo-like kinase (NLK)........................................................................ 23 Regulation of NLK expression..................................................... 23 The subcellular localization of NLK............................................. 24 Mechanism of activation of NLK.................................................. 24 Physiological functions of NLK.................................................... 25 NLK as a regulator of signaling pathways................................... 26 NLK and disease......................................................................... 30 Wnt signaling pathway......................................................................... 31 Wnt ligands, biogenesis and extracellular transport................... 32 Receptors dictate Wnt activity..................................................... 33 Wnt--catenin signaling............................................................... 34 Objectives................................................................................................ 39 Materials and Methods............................................................................ 43 1. Plasmids............................................................................................ 45 1.1. Backbone vectors................................................................. 45 1.2. DYRK1A expression vectors................................................ 45 1.3. Other DYRKs expression vectors........................................ 46 1.4. NLK expression vectors....................................................... 46 1.5. -catenin expression vectors............................................... 46 1.6. Other plasmids..................................................................... 47 2. Techniques for DNA manipulation................................................... 48 2.1. DNA purification and sequencing......................................... 48 2.2. Cloning of a DNA fragment into an expression vector......... 48 2.3. Site-directed mutagenesis.................................................... 50 3. Cell culture........................................................................................ 51 3.1. Cell lines............................................................................... 51 3.2. Cell transfection.................................................................... 51 3.3. Cell treatments..................................................................... 51 3.4. Pulse-chase experiments..................................................... 52 3.5. Preparation of lentivirus stocks and infection....................... 52 4. Techniques for protein analysis........................................................ 53 4.1. Preparation of cell lysates.................................................... 53 4.2. Western blot analysis........................................................... 53 4.3. Immunoprecipitation assays................................................. 54 4.4. Immunofluorescence............................................................ 55 4.5. Purification of GST-fusion proteins....................................... 55 4.6. Pull-down assays................................................................. 56 4.7. In vitro kinase assays........................................................... 57 4.8. Mass spectrometry analysis................................................. 57 4.9. Reporter assays................................................................... 58 4.10. Protein quantification.......................................................... 58 5. Bioinformatic tools and statistical analysis....................................... 58 5.1. Search in the databases....................................................... 58 5.2. DNA sequence analysis....................................................... 59 5.3. Protein analysis.................................................................... 59 5.4. Statistical analysis................................................................ 59 Results..................................................................................................... 63 DYRK1A interacts with NLK................................................................. 65 The interaction between DYRK1A and NLK is independent of their kinase activities.................................................................................... 66 NLK interacts with all DYRK family members....................................... 67 The N-terminal region of DYRK1A has an NLK-binding site................ 68 NLK interacts with DYRK1A through its C-terminal region.................. 71 NLK phosphorylates DYRK1A............................................................. 73 NLK phosphorylation of DYRK1A affects neither enzymatic activity nor subcellular localization................................................................... 73 NLK co-expression induces a decrease in the accumulation of DYRK1A............................................................................................... 76 NLK induces DYRK1A proteasome-dependent degradation................ 77 NLK affects DYRK1A half-life through phosphorylation of multiple sites...................................................................................................... 79 The Wnt1/Wnt3a-Fzd1-HIPK2-NLK signaling cascade affects DYRK1A half-life................................................................................... 82 DYRK1A is a positive regulator of the transcriptional activity of a - catenin-dependent reporter.................................................................. 83 DYRK1A interacts with -catenin......................................................... 85 DYRK1A forms a ternary complex with the -catenin/TCF transcriptional complex......................................................................... 86 DYRK1A phosphorylates -catenin at multiple sites............................ 87 DYRK1A enhances -catenin-dependent transcriptional activity through phosphorylation of serine residues 552 and 675..................... 90 NLK inhibits the DYRK1A enhancement on the -catenin-dependent transcriptional activity........................................................................... 91 Discussion............................................................................................... 93 The protein kinases DYRK1A and NLK interact................................... 95 DYRK1A is a substrate of NLK............................................................. 98 DYRK1A plays a positive role on -catenin-dependent transcriptional activity................................................................................................... 101 Final remarks and future prospects...................................................... 105 Conclusions............................................................................................. 109 References............................................................................................... 113 Abbreviations.......................................................................................... 131 ABSTRACT Abstract DYRK1A is the most studied member of the DYRK family of protein kinases, because is one of the human chromosoma 21 proteins for which changes in gene dosage result in neuropathological alterations. DYRKs are activated by autophosphorylation on a tyrosine residue in the activation loop, a one-off event that takes place during translation. Accordingly, DYRK1A would be constitutively active once is synthesized. However, DYRK1A is extremely sensitive to gene dosage, and thus it is predictable that not only
Recommended publications
  • Aberrant Methylation Underlies Insulin Gene Expression in Human Insulinoma
    ARTICLE https://doi.org/10.1038/s41467-020-18839-1 OPEN Aberrant methylation underlies insulin gene expression in human insulinoma Esra Karakose1,6, Huan Wang 2,6, William Inabnet1, Rajesh V. Thakker 3, Steven Libutti4, Gustavo Fernandez-Ranvier 1, Hyunsuk Suh1, Mark Stevenson 3, Yayoi Kinoshita1, Michael Donovan1, Yevgeniy Antipin1,2, Yan Li5, Xiaoxiao Liu 5, Fulai Jin 5, Peng Wang 1, Andrew Uzilov 1,2, ✉ Carmen Argmann 1, Eric E. Schadt 1,2, Andrew F. Stewart 1,7 , Donald K. Scott 1,7 & Luca Lambertini 1,6 1234567890():,; Human insulinomas are rare, benign, slowly proliferating, insulin-producing beta cell tumors that provide a molecular “recipe” or “roadmap” for pathways that control human beta cell regeneration. An earlier study revealed abnormal methylation in the imprinted p15.5-p15.4 region of chromosome 11, known to be abnormally methylated in another disorder of expanded beta cell mass and function: the focal variant of congenital hyperinsulinism. Here, we compare deep DNA methylome sequencing on 19 human insulinomas, and five sets of normal beta cells. We find a remarkably consistent, abnormal methylation pattern in insu- linomas. The findings suggest that abnormal insulin (INS) promoter methylation and altered transcription factor expression create alternative drivers of INS expression, replacing cano- nical PDX1-driven beta cell specification with a pathological, looping, distal enhancer-based form of transcriptional regulation. Finally, NFaT transcription factors, rather than the cano- nical PDX1 enhancer complex, are predicted to drive INS transactivation. 1 From the Diabetes Obesity and Metabolism Institute, The Department of Surgery, The Department of Pathology, The Department of Genetics and Genomics Sciences and The Institute for Genomics and Multiscale Biology, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
    [Show full text]
  • Correction of Cognitive Deficits in Mouse Models Of
    Correction of cognitive deficits in mouse models of Down syndrome by a pharmacological inhibitor of DYRK1A Thu Lan Nguyen, Arnaud Duchon, Antigoni Manousopoulou, Nadège Loaëc, Benoît Villiers, Guillaume Pani, Meltem Karatas, Anna E Mechling, Laura-Adela Harsan, Emmanuelle Limanton, et al. To cite this version: Thu Lan Nguyen, Arnaud Duchon, Antigoni Manousopoulou, Nadège Loaëc, Benoît Villiers, et al.. Correction of cognitive deficits in mouse models of Down syndrome by a pharmacological in- hibitor of DYRK1A. Disease Models & Mechanisms, Cambridge Company of Biologists, 2018, 11 (9), pp.dmm035634. 10.1242/dmm.035634. hal-01862465 HAL Id: hal-01862465 https://hal-univ-rennes1.archives-ouvertes.fr/hal-01862465 Submitted on 17 Jul 2019 HAL is a multi-disciplinary open access L’archive ouverte pluridisciplinaire HAL, est archive for the deposit and dissemination of sci- destinée au dépôt et à la diffusion de documents entific research documents, whether they are pub- scientifiques de niveau recherche, publiés ou non, lished or not. The documents may come from émanant des établissements d’enseignement et de teaching and research institutions in France or recherche français ou étrangers, des laboratoires abroad, or from public or private research centers. publics ou privés. © 2018. Published by The Company of Biologists Ltd | Disease Models & Mechanisms (2018) 11, dmm035634. doi:10.1242/dmm.035634 RESEARCH ARTICLE Correction of cognitive deficits in mouse models of Down syndrome by a pharmacological inhibitor of DYRK1A Thu Lan Nguyen1,2,3,4,5, Arnaud Duchon1,2,3,4, Antigoni Manousopoulou6, Nadegè Loaëc5, Benoît Villiers5, Guillaume Pani1,2,3,4, Meltem Karatas7,8, Anna E. Mechling8, Laura-Adela Harsan7,8, Emmanuelle Limanton9, Jean-Pierre Bazureau9, François Carreaux9, Spiros D.
    [Show full text]
  • The Mutational Landscape of Myeloid Leukaemia in Down Syndrome
    cancers Review The Mutational Landscape of Myeloid Leukaemia in Down Syndrome Carini Picardi Morais de Castro 1, Maria Cadefau 1,2 and Sergi Cuartero 1,2,* 1 Josep Carreras Leukaemia Research Institute (IJC), Campus Can Ruti, 08916 Badalona, Spain; [email protected] (C.P.M.d.C); [email protected] (M.C.) 2 Germans Trias i Pujol Research Institute (IGTP), Campus Can Ruti, 08916 Badalona, Spain * Correspondence: [email protected] Simple Summary: Leukaemia occurs when specific mutations promote aberrant transcriptional and proliferation programs, which drive uncontrolled cell division and inhibit the cell’s capacity to differentiate. In this review, we summarize the most frequent genetic lesions found in myeloid leukaemia of Down syndrome, a rare paediatric leukaemia specific to individuals with trisomy 21. The evolution of this disease follows a well-defined sequence of events and represents a unique model to understand how the ordered acquisition of mutations drives malignancy. Abstract: Children with Down syndrome (DS) are particularly prone to haematopoietic disorders. Paediatric myeloid malignancies in DS occur at an unusually high frequency and generally follow a well-defined stepwise clinical evolution. First, the acquisition of mutations in the GATA1 transcription factor gives rise to a transient myeloproliferative disorder (TMD) in DS newborns. While this condition spontaneously resolves in most cases, some clones can acquire additional mutations, which trigger myeloid leukaemia of Down syndrome (ML-DS). These secondary mutations are predominantly found in chromatin and epigenetic regulators—such as cohesin, CTCF or EZH2—and Citation: de Castro, C.P.M.; Cadefau, in signalling mediators of the JAK/STAT and RAS pathways.
    [Show full text]
  • Eif2b Is a Decameric Guanine Nucleotide Exchange Factor with a G2e2 Tetrameric Core
    ARTICLE Received 19 Feb 2014 | Accepted 15 Apr 2014 | Published 23 May 2014 DOI: 10.1038/ncomms4902 OPEN eIF2B is a decameric guanine nucleotide exchange factor with a g2e2 tetrameric core Yuliya Gordiyenko1,2,*, Carla Schmidt1,*, Martin D. Jennings3, Dijana Matak-Vinkovic4, Graham D. Pavitt3 & Carol V. Robinson1 eIF2B facilitates and controls protein synthesis in eukaryotes by mediating guanine nucleotide exchange on its partner eIF2. We combined mass spectrometry (MS) with chemical cross- linking, surface accessibility measurements and homology modelling to define subunit stoichiometry and interactions within eIF2B and eIF2. Although it is generally accepted that eIF2B is a pentamer of five non-identical subunits (a–e), here we show that eIF2B is a decamer. MS and cross-linking of eIF2B complexes allows us to propose a model for the subunit arrangements within eIF2B where the subunit assembly occurs through catalytic g- and e-subunits, with regulatory subunits arranged in asymmetric trimers associated with the core. Cross-links between eIF2 and eIF2B allow modelling of interactions that contribute to nucleotide exchange and its control by eIF2 phosphorylation. Finally, we identify that GTP binds to eIF2Bg, prompting us to propose a multi-step mechanism for nucleotide exchange. 1 Department of Chemistry, Physical and Theoretical Chemistry Laboratory, University of Oxford, South Parks Road, Oxford OX1 3QZ, UK. 2 MRC Laboratory of Molecular Biology, University of Cambridge, Francis Crick Avenue, Cambridge CB2 0QH, UK. 3 Faculty of Life Sciences, Michael Smith Building, University of Manchester, Oxford Road, Manchester M13 9PT, UK. 4 Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, UK.
    [Show full text]
  • Strategies and Opportunities for Small Molecule Drug Discovery to Target Neurodegenerative Diseases Andrea I
    bioRxiv preprint doi: https://doi.org/10.1101/2020.04.01.020206; this version posted April 2, 2020. The copyright holder has placed this preprint (which was not certified by peer review) in the Public Domain. It is no longer restricted by copyright. Anyone can legally share, reuse, remix, or adapt this material for any purpose without crediting the original authors. Defining the Neural Kinome: Strategies and Opportunities for Small Molecule Drug Discovery to Target Neurodegenerative Diseases Andrea I. Krahn, Carrow Wells, David H. Drewry, Lenore K. Beitel, Thomas M. Durcan, Alison D. Axtman* ABSTRACT: Kinases are highly tractable drug targets that have reached unparalleled success in fields such as cancer but whose potential has not yet been realized in neuroscience. There are currently 55 approved small molecule kinase-targeting drugs, 48 of which have an anti-cancer indication. The intrinsic complexity linked to central nervous system (CNS) drug development and a lack of validated targets has hindered progress in developing kinase inhibitors for CNS disorders when compared to other therapeutic areas such as oncology. Identification and/or characterization of new kinases as potential drug targets for neurodegenerative diseases will create opportunities for development of CNS drugs in the future. The track record of kinase inhibitors in other disease indications supports the idea that with the best targets identified small molecule kinase modulators will become impactful therapeutics for neurodegenerative diseases. KEYWORDS: kinase, neurodegeneration,
    [Show full text]
  • DYRK1A Binds to an Evolutionarily Conserved WD40-Repeat Title Protein WDR68 and Induces Its Nuclear Translocation
    DYRK1A binds to an evolutionarily conserved WD40-repeat Title protein WDR68 and induces its nuclear translocation. Author(s) Miyata, Yoshihiko; Nishida, Eisuke Citation Biochimica et biophysica acta (2011), 1813(10): 1728-1739 Issue Date 2011-10 URL http://hdl.handle.net/2433/148020 © 2011 Elsevier B.V.; This is not the published version. Please cite only the published version.; この論文は出版社版であり Right ません。引用の際には出版社版をご確認ご利用ください 。 Type Journal Article Textversion author Kyoto University *REVISED Manuscript (text UNmarked) Click here to view linked References DYRK1A binds to an evolutionarily conserved WD40-repeat protein WDR68 and induces its nuclear translocation Yoshihiko Miyata*, Eisuke Nishida Department of Cell and Developmental Biology, Graduate School of Biostudies, Kyoto University, Kitashirakawa Oiwake-cho, Kyoto 606-8502, Japan * Corresponding author. Department of Cell & Developmental Biology, Graduate School of Biostudies, Kyoto University, Kitashirakawa Oiwake-cho, Sakyo-ku, Kyoto 606-8502, Japan. Tel.: +81-75-753-4231; fax: +81-75-753-4235. 1 ABSTRACT DYRK1A is encoded in the Down’s syndrome critical region on human chromosome 21, and plays an important role in the functional and developmental regulation of many types of cells, including neuronal cells. Here we have identified WDR68, an evolutionarily conserved protein with WD40-repeat domains, as a cellular binding partner of DYRK1A. WDR68 was originally identified in petunia as AN11 that controls the pigmentation of flowers by stimulating the transcription of anthocyanin biosynthetic genes. Experiments with RNA interference showed that WDR68 was indispensable for the optimal proliferation and survival of mammalian cultured cell, and WDR68 depletion induced cell apoptosis. DYRK1A and DYRK1B, but not DYRK2, DYRK3, or DYRK4, bound to endogenous and expressed WDR68.
    [Show full text]
  • Cofactor Dependent Conformational Switching of Gtpases
    View metadata, citation and similar papers at core.ac.uk brought to you by CORE provided by Elsevier - Publisher Connector 1704 Biophysical Journal Volume 95 August 2008 1704–1715 Cofactor Dependent Conformational Switching of GTPases Vasili Hauryliuk,*y Sebastian Hansson,z and Ma˚ns Ehrenberg* *Department of Cell and Molecular Biology, Molecular Biology Program, Uppsala University, Uppsala, Sweden; yEngelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia; and zLaboratoire d’Enzymologie et Biochimie Structurales, CNRS, Avenue de la Terrasse, 91198 Gif-sur-Yvette, France ABSTRACT This theoretical work covers structural and biochemical aspects of nucleotide binding and GDP/GTP exchange of GTP hydrolases belonging to the family of small GTPases. Current models of GDP/GTP exchange regulation are often based on two specific assumptions. The first is that the conformation of a GTPase is switched by the exchange of the bound nucleotide from GDP to GTP or vice versa. The second is that GDP/GTP exchange is regulated by a guanine nucleotide exchange factor, which stabilizes a GTPase conformation with low nucleotide affinity. Since, however, recent biochemical and structural data seem to contradict this view, we present a generalized scheme for GTPase action. This novel ansatz accounts for those important cases when conformational switching in addition to guanine nucleotide exchange requires the presence of cofactors, and gives a more nuanced picture of how the nucleotide exchange is regulated. The scheme is also used to discuss some problems of interpretation that may arise when guanine nucleotide exchange mechanisms are inferred from experiments with analogs of GTP, like GDPNP, GDPCP, and GDP g S.
    [Show full text]
  • DYRK1A Inhibition and Cognitive Rescue in a Down Syndrome Mouse
    www.nature.com/scientificreports OPEN DYRK1A inhibition and cognitive rescue in a Down syndrome mouse model are induced by new fuoro- Received: 15 June 2017 Accepted: 18 January 2018 DANDY derivatives Published: xx xx xxxx Fernanda Neumann1, Stéphanie Gourdain1, Christelle Albac2, Alain D. Dekker 2,3, Linh Chi Bui4, Julien Dairou5, Isabelle Schmitz-Afonso1, Nathalie Hue1, Fernando Rodrigues-Lima4, Jean M. Delabar2, Marie-Claude Potier2, Jean-Pierre Le Caër1, David Touboul1, Benoît Delatour2, Kevin Cariou1 & Robert H. Dodd1 Inhibition of DYRK1A kinase, produced by chromosome 21 and consequently overproduced in trisomy 21 subjects, has been suggested as a therapeutic approach to treating the cognitive defciencies observed in Down syndrome (DS). We now report the synthesis and potent DYRK1A inhibitory activities of fuoro derivatives of 3,5-di(polyhydroxyaryl)-7-azaindoles (F-DANDYs). One of these compounds (3-(4-fuorophenyl)-5-(3,4-dihydroxyphenyl)-1H-pyrrolo[2,3-b]pyridine, 5a) was selected for in vivo studies of cognitive rescuing efects in a standard mouse model of DS (Ts65Dn line). Using the Morris water maze task, Ts65Dn mice treated i.p. with 20 mg/kg of 5a performed signifcantly better than Ts65Dn mice treated with placebo, confrming the promnesiant efect of 5a in the trisomic mice. Overall, these results demonstrate for the frst time that selective and competitive inhibition of DYRK1A kinase by the F-DANDY derivative 5a may provide a viable treatment strategy for combating the memory and learning defciencies encountered in DS. Down syndrome (DS), or trisomy 21, is the most common genetically acquired form of intellectual disability1–3, occurring in approximately 1 out of 650–1000 newborns in Europe4,5 and North America6.
    [Show full text]
  • A Dissertation Entitled Characterization of the CXCR4
    A Dissertation entitled Characterization of the CXCR4-LASP1-eIF4F Axis in Triple-Negative Breast Cancer by Cory M Howard Submitted to the Graduate Faculty as partial fulfillment of the requirements for the Doctor of Philosophy Degree in Biomedical Sciences ___________________________________________ Dayanidhi Raman, B.V.Sc., Ph.D., Committee Chair ___________________________________________ Amit Tiwari, Ph.D., Committee Member ___________________________________________ Ritu Chakravarti, Ph.D., Committee Member ___________________________________________ Nagalakshmi Nadiminty, Ph.D., Committee Member ___________________________________________ Saori Furuta, Ph.D., Committee Member ___________________________________________ Shi-He Liu, M.D., Committee Member ___________________________________________ Amanda C. Bryant-Friedrich, Ph.D., Dean College of Graduate Studies The University of Toledo August 2020 © 2020 Cory M. Howard This document is copyrighted material. Under copyright law, no parts of this document may be reproduced without the expressed permission of the author. An Abstract of Characterization of the CXCR4-LASP1-eIF4F Axis in Triple-Negative Breast Cancer by Cory M. Howard Submitted to the Graduate Faculty as partial fulfillment of the requirements for the Doctor of Philosophy Degree in Biomedical Sciences The University of Toledo August 2020 Triple-negative breast cancer (TNBC) remains clinically challenging as effective targeted therapies are still lacking. In addition, patient mortality mainly results from the metastasized
    [Show full text]
  • Druggable Transient Pockets in Protein Kinases
    molecules Review Druggable Transient Pockets in Protein Kinases Koji Umezawa 1 and Isao Kii 2,* 1 Department of Biomolecular Innovation, Institute for Biomedical Sciences, Shinshu University, 8304 Minami-Minowa, Kami-ina, Nagano 399-4598, Japan; [email protected] 2 Laboratory for Drug Target Research, Faculty & Graduate School of Agriculture, Shinshu University, 8304 Minami-Minowa, Kami-ina, Nagano 399-4598, Japan * Correspondence: [email protected]; Tel.: +81-265-77-1521 Abstract: Drug discovery using small molecule inhibitors is reaching a stalemate due to low se- lectivity, adverse off-target effects and inevitable failures in clinical trials. Conventional chemical screening methods may miss potent small molecules because of their use of simple but outdated kits composed of recombinant enzyme proteins. Non-canonical inhibitors targeting a hidden pocket in a protein have received considerable research attention. Kii and colleagues identified an inhibitor targeting a transient pocket in the kinase DYRK1A during its folding process and termed it FINDY. FINDY exhibits a unique inhibitory profile; that is, FINDY does not inhibit the fully folded form of DYRK1A, indicating that the FINDY-binding pocket is hidden in the folded form. This intriguing pocket opens during the folding process and then closes upon completion of folding. In this review, we discuss previously established kinase inhibitors and their inhibitory mechanisms in comparison with FINDY. We also compare the inhibitory mechanisms with the growing concept of “cryptic inhibitor-binding sites.” These sites are buried on the inhibitor-unbound surface but become apparent when the inhibitor is bound. In addition, an alternative method based on cell-free protein synthesis of protein kinases may allow the discovery of small molecules that occupy these mysterious binding sites.
    [Show full text]
  • Nck Adapter Proteins: Functional Versatility in T Cells Marcus Lettau, Jennifer Pieper and Ottmar Janssen
    Cell Communication and Signaling BioMed Central Review Open Access Nck adapter proteins: functional versatility in T cells Marcus Lettau, Jennifer Pieper and Ottmar Janssen Address: 1University Hospital Schleswig-Holstein Campus Kiel, Institute of Immunology, Molecular Immunology, Arnold-Heller-Str 3, Bldg 17, D-24105 Kiel, Germany E-mail: Marcus Lettau* - [email protected]; Jennifer Pieper - [email protected]; Ottmar Janssen - [email protected] *Corresponding author Published: 02 February 2009 Received: 2 December 2008 Cell Communication and Signaling 2009, 7:1 doi: 10.1186/1478-811X-7-1 Accepted: 2 February 2009 This article is available from: http://www.biosignaling.com/content/7/1/1 © 2009 Lettau et al; licensee BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Abstract Nck is a ubiquitously expressed adapter protein that is almost exclusively built of one SH2 domain and three SH3 domains. The two isoproteins of Nck are functionally redundant in many aspects and differ in only few amino acids that are mostly located in the linker regions between the interaction modules. Nck proteins connect receptor and non-receptor tyrosine kinases to the machinery of actin reorganisation. Thereby, Nck regulates activation-dependent processes during cell polarisation and migration and plays a crucial role in the signal transduction of a variety of receptors including for instance PDGF-, HGF-, VEGF- and Ephrin receptors. In most cases, the SH2 domain mediates binding to the phosphorylated receptor or associated phosphoproteins, while SH3domaininteractionsleadtotheformationoflargerproteincomplexes.InTlymphocytes,Nck plays a pivotal role in the T cell receptor (TCR)-induced reorganisation of the actin cytoskeleton and the formation of the immunological synapse.
    [Show full text]
  • Buy Viagra Brand
    A Canonical Cancer-Network Map Eugene F. Douglass Jr., Ph.D. ;<=:>&'? Cell-Surface Interactions @@@JJ; Growth Receptor Signalling Cell-ECM Growth/Motility Proliferation Cell-Cell Contact Inhibition Metabolism MMP COLONIC CRYPT IGF/IGFR-1 EGFR / ErbB WNT ! WNT ! WNT WNT Integrin WNT RAS must be held near PI3K to activate it 5 CAD-domains Signalosome R-spdn p.195 GTP RAS PTEN NF1 SOS PLC PIP3 E-cadherin SHC PIP RASGAP GRB2 2 ZNRF3 p120 p120 " p120 p120 p120 p120 FZD Akt PDK1 PH JM Src-dep Y845 STAT3 GDP GTP Y974 F-actin PI3K PTB SHC/SHP2 GRB2 LRP Assembly vincilin talin PI3K Integrin-Linked Kinase(ILK) Y981 RAS RAS LGR RNF43 NCK2 ! ! ! ARP2/3 FAK Y988 FAK 2 min PINCH SOCS3 C hevell Akt KD s e paxilin SOS i d PDK1 Src IRS1 ! ! ! d paxilin Src (PKB) internlz AP2 RSU1 Y1015 ½ HIC5 ! t PI3K Y460 SOCS1/6 Y992 PLC" Parvin Src TESK1 PI3K Y546 SHIP2 GTP p130Cas CRK CSG ubiq axin PAK1 Y1091 Y1045 CBL S46-7 RAF-1 KSR DOCK180 SOS PI3K Y608 S1057 = GSK3 PI3K Y628 Y1127 Y1068 GRB2 14-3-3 14-3-3 PI3K Y658 GRB2 Y1086 B-RAF C-RAF GSK RHEB PI3K Y690 RASGAP 9 Wtx mTOR1 Y1163 Y1101 Gab1 Apc PI3K Y727 Y1167 PI3K Y759 RASGAP Y1168 MAPK Rac GTP Y1148 SHC GRB2 p120 p120 Y815 SHIP2 RhoGEF RalGEF p70-S6K Y1215 ! ! Y1175 SOS GTP Y891 phos SHP1 Cdc42 Y903 SHIP2 Y1253 SHP2 GRB2 ! ! PTP1 MKK1/2 PI3K Y935 Wtx GTP CT ! Rho PI3K Y983 SHIP2 Y1282 GRB2 SHP2 Y1006 Y1283 RalBP1 RalA/B GTP MP1 axin PI3K Y1352 PI3K Erk1/2 GSK3 Sprouty Apc MKP1 GTP GRB2 SHP2 Y1173 Rac 1.
    [Show full text]